2k Clinical Consulting, Inc.

From Stress to Success: Coping Strategies for Clinical Site Staff During FDA Inspections

FDA inspections, while critical for ensuring the safety and efficacy of clinical research, can be a significant source of stress for clinical site staff. The anticipation and the process itself can create anxiety, but with the right strategies, your team can navigate these challenges successfully and even use the experience as an opportunity for professional growth and improvement. Here are some effective ways to help clinical site staff cope with the pressures of FDA inspections and turn stressful situations into success stories.

Preparation is the Best Defense

Thorough preparation is the most effective way to reduce stress during FDA inspections. Ensuring that all staff are well-trained and understand their roles and responsibilities is crucial. This includes regular reviews of GCP guidelines, internal SOPs, and any recent updates to FDA regulations. Conduct mock inspections to familiarize staff with the inspection process. This not only helps identify any gaps in compliance but also boosts confidence across the team.

Maintain Open Lines of Communication

Create an environment where staff feel comfortable communicating their concerns and questions. Prior to an inspection, hold briefings to go over what to expect and encourage team members to share their experiences and strategies for handling potential stress points. During the inspection, maintain open lines of communication to manage any unexpected issues swiftly and efficiently.

Support System and Teamwork

Encourage a strong support network within your team. Stress can be significantly alleviated when staff feel they are part of a cohesive unit working towards a common goal. Foster a team-oriented atmosphere where everyone knows they have the support of their colleagues and superiors. This includes having clear, accessible points of contact for when they need guidance or reassurance during the inspection process.

Physical and Mental Preparedness

Remind staff of the importance of personal well-being during stressful times. Encourage regular breaks, proper hydration, and, if possible, provide a quiet space for relaxation during lengthy inspection days. Mental preparedness is just as important, and practices such as mindfulness or stress-reduction techniques can be beneficial.

Post-Inspection Debrief and Continuous Improvement

After the inspection, it’s important to gather the team for a debrief session. Discuss what went well and what could be improved for future inspections. This not only helps in identifying areas for development but also allows the team to collectively process the experience and relieve post-inspection stress. Implementing changes based on feedback demonstrates a commitment to continuous improvement and helps build confidence in handling future inspections.

Encouraging Professional Development

Invest in the professional development of your staff. Provide opportunities for further training and education in areas highlighted during inspections. This not only improves compliance and performance but also empowers staff by giving them the tools they need to succeed.

By turning the stress of FDA inspections into a constructive force, clinical site staff can improve their professional capabilities and enhance the overall quality of the research conducted. With the right preparation, support, and continuous improvement, your team can transform potential challenges into achievements, maintaining compliance and ensuring successful outcomes.

To further enhance your team’s ability to manage the stress of FDA inspections and to foster a proactive, knowledgeable workplace, consider enrolling in specialized courses such as “Preparing for FDA GCP Inspections – Best Practices for Clinical Sites.” This training can provide deeper insights and more structured approaches to not only surviving but thriving during FDA inspections. Enroll in the course and take a significant step towards empowering your clinical site staff.

Navigating Common FDA Inspection Challenges: Empowering Clinical Sites

FDA inspections are a critical aspect of clinical research, ensuring adherence to GCP standards and safeguarding participant well-being. Our comprehensive guide is designed to empower your clinical site to navigate these inspections successfully, transforming potential challenges into opportunities for enhancement and compliance.

Anticipating and Addressing Frequent Pitfalls

Identifying and addressing common pitfalls is a key step in preparing for an FDA inspection. Frequent issues include inadequate informed consent documentation, non-adherence to protocols, and inaccurate record-keeping. By proactively tackling these issues, your site can demonstrate a strong commitment to regulatory compliance and patient safety. Regular internal audits and pre-inspection checks are effective strategies for mitigating these risks before an inspection occurs.

Empowering Your Team with Knowledge and Resources

The readiness of your team is crucial to the success of an FDA inspection. Providing regular training on GCP guidelines and FDA-specific requirements equips your staff with essential knowledge. Establishing clear, documented protocols for maintaining and documenting compliance is also critical. Investing in continuous education and resources fosters a culture where quality and compliance are paramount.

Engaging in Continuous Improvement

Viewing FDA inspections as opportunities for improvement rather than merely evaluative exercises can significantly change your team’s approach to compliance. Implementing a systematic review process post-inspection allows your team to address any identified deficiencies and continuously refine and enhance operational practices. This ongoing improvement ensures that your site is always prepared for inspections.

Leveraging Expert Guidance

For those looking to deepen their understanding and enhance their inspection readiness, the course “Preparing for FDA GCP Inspections – Best Practices for Clinical Sites” provides comprehensive insights into preparing for and navigating through FDA inspections. This course offers practical, actionable strategies that can be implemented to enhance your site’s preparedness.

Conclusion

Maintaining compliance and readiness for FDA inspections is achievable with the right strategies, continuous improvement, and access to expert guidance. By embracing these practices, your clinical site can approach inspections with confidence and demonstrate a strong commitment to research integrity and participant safety.

For a more detailed exploration of best practices in FDA inspection readiness, consider enrolling in “Preparing for FDA GCP Inspections – Best Practices for Clinical Sites.” This course is designed to provide you with the knowledge and tools needed to excel in the face of regulatory scrutiny.   Visit our website and enroll in the course today. Ensure your clinical site exceeds the standards set by regulatory authorities.

Elevating Clinical Site Management: Key Strategies for GCP Compliance

Clinical site management is a pivotal aspect of clinical research, ensuring the integrity, safety, and efficacy of trials. Adherence to Good Clinical Practice (GCP) is non-negotiable, demanding meticulous attention and robust strategies. By elevating site management practices, we can achieve and maintain GCP compliance, enhancing the quality of our research and the safety of its participants.

Comprehensive Training and Education

Education is the bedrock of effective site management. Investing in comprehensive, ongoing training for site staff ensures a deep understanding of GCP requirements and how they translate into daily operations. Training should be dynamic, incorporating the latest regulatory updates and best practices, and should engage staff at all levels, fostering a culture of continuous learning and adherence.

Engagement in Continuous Professional Development:

  • Regularly updated GCP training programs.
  • Inclusion of real-world scenarios and problem-solving exercises.
  • Assessment and certification to validate understanding and commitment.

Robust Process and Documentation

GCP compliance hinges on rigorous processes and thorough documentation, establishing a clear, auditable trail of every aspect of the trial. Implementing standardized procedures across sites ensures consistency and quality, while meticulous documentation demonstrates compliance and facilitates effective oversight.

Key Elements of Process and Documentation:

  • Standard Operating Procedures (SOPs) tailored to GCP guidelines.
  • Rigorous record-keeping and data integrity measures.
  • Regular audits to ensure compliance and identify areas for improvement.

Effective Communication and Collaboration

Transparent and effective communication between site staff, sponsors, and regulators is crucial for GCP compliance. Encouraging open dialogue and collaboration fosters a proactive approach to compliance, enabling swift resolution of potential issues and continuous improvement of site practices.

Strategies for Enhancing Communication:

  • Regular meetings and updates between all stakeholders.
  • Clear channels for raising concerns and reporting issues.
  • Collaborative problem-solving and decision-making processes.

Leveraging Technology

Innovative technologies can streamline site management and enhance GCP compliance. From electronic data capture (EDC) systems to advanced monitoring tools, technology can reduce errors, improve efficiency, and provide real-time insights into compliance status.

Technology-Driven Improvements:

  • Adoption of EDC and Clinical Trial Management Systems (CTMS).
  • Utilization of remote monitoring and telemedicine where appropriate.
  • Investment in training to ensure proficient use of technological tools.

Continuous Improvement and Feedback

The landscape of clinical research is ever-evolving, and so are the standards of GCP. Establishing mechanisms for feedback, review, and continuous improvement ensures that site management practices remain at the forefront of quality and compliance.

Cultivating a Culture of Improvement:

  • Regular feedback sessions with site staff and stakeholders.
  • Implementation of lessons learned from audits and inspections.
  • Commitment to evolving practices in line with regulatory changes and industry advancements.

Elevating clinical site management is a collaborative, ongoing effort that requires commitment from all stakeholders. By embracing these strategies, we can uphold the highest standards of GCP compliance, ensuring the success and integrity of clinical research.

We value your insights and experiences in this critical area. How do you ensure GCP compliance in your clinical site management? 

Through collective effort and shared commitment, we can continue to advance the field of clinical research, ensuring it is conducted to the highest standards of safety, ethics, and scientific integrity.

Enhancing Compliance: Aligning Vendor Practices with FDA Requirements 

In the rapidly evolving landscape of healthcare and pharmaceuticals, sponsor companies face the critical challenge of ensuring that their vendor practices align seamlessly with FDA requirements. This alignment is not just a regulatory mandate but a cornerstone for maintaining trust, ensuring product safety, and fostering innovation. By delving into the strategies for achieving this alignment, companies can navigate the complexities of compliance while sustaining growth and market reputation. 

Understanding FDA Requirements – The first step toward alignment is a thorough understanding of FDA regulations, which encompass a wide array of areas including product development, clinical trials, manufacturing, and post-market surveillance. These regulations are designed to ensure that products are safe, effective, and of high quality. For sponsor companies, this means ensuring that every vendor, from raw material suppliers to contract research organizations, adheres to these stringent standards. 

Strategic Vendor Selection – The cornerstone of compliance is the meticulous selection of vendors. Sponsor companies must conduct due diligence to assess a vendor’s capability to meet FDA requirements. This includes evaluating their quality control systems, regulatory history, and operational capacity. By choosing vendors with a robust track record of compliance, companies can mitigate risks and foster a culture of quality and reliability. 

Fostering Collaboration and Transparency – Building a transparent and collaborative relationship with vendors is crucial for compliance. This involves clear communication of expectations, regular audits, and continuous feedback mechanisms. By fostering an environment where information and best practices are shared openly, sponsor companies and vendors can work together to identify potential compliance issues and implement corrective actions proactively. 

Implementing Robust Monitoring and Auditing – Ongoing monitoring and auditing of vendor practices are essential to ensure continuous alignment with FDA requirements. This includes regular inspections, performance reviews, and the tracking of compliance metrics. By establishing a systematic approach to monitoring, sponsor companies can promptly address any deviations and implement improvements, thereby minimizing compliance risks. 

Leveraging Technology for Compliance – In today’s digital age, leveraging technology can significantly enhance compliance efforts. Advanced software solutions can streamline compliance monitoring, facilitate data sharing between sponsors and vendors, and provide real-time insights into vendor performance. By harnessing technology, sponsor companies can achieve greater visibility and control over their supply chain, ensuring that every link aligns with FDA standards. 

In conclusion, aligning vendor practices with FDA requirements is a dynamic and ongoing process that demands diligence, collaboration, and innovation. By understanding the regulatory landscape, selecting the right partners, fostering transparency, and leveraging technology, sponsor companies can establish a robust compliance framework. This not only safeguards product integrity and patient safety but also enhances the company’s reputation and market success. 

Your Feedback Matters How is your organization aligning vendor practices with FDA requirements? Are there specific challenges or success stories you would like to share? Your insights are invaluable, and we invite you to share your experiences and feedback to foster a collaborative learning community. 

By engaging with these strategies and sharing insights, sponsor companies can not only comply with regulatory mandates but also pave the way for a future where safety, quality, and innovation go hand in hand. 

Steps To Establishing a Quality Culture in Clinical Research

 

A quality culture pertains to the shared beliefs, values, and attitudes that incorporates a quality mindset which also influences the actions taken by individuals within an organization. It is essential to establish a quality culture in the workplace in clinical trials to ensure high-quality data is collected and patient safety is maintained.  Establishing a quality culture in clinical research is not just about having policies and procedures in place, it is also about ensuring that everyone follows them. This requires implementing several important steps.

Step One: Define quality expectations

The first step in establishing a quality culture is to identify the quality standards, processes, procedures, and timelines that must be followed to ensure high-quality research. It is required to develop clear expectations for data collection, monitoring, and reporting, as well as define requirements for maintaining compliance with regulatory requirements and industry standards. Make sure that everyone involved in the research understands what is expected of them in terms of quality and patient safety.

Step Two:  Develop quality metrics

The next step is to create a set of measurable indicators that can be used to monitor and evaluate the effectiveness of the quality management system. These metrics should be clearly defined and aligned with the organization’s quality expectations. Examples of quality metrics in clinical trials may include the number of protocol deviations, the percentage of completed monitoring visits, and the number of audit findings.

Step Three: Establish a quality management system

Establishing and implementing a clinical quality management system involves creating a set of policies, procedures, and practices that ensure that all processes are in place to achieve quality expectations. It is crucial to develop standard operating procedures (SOPs) for data collection, analysis, and reporting, as well as implement a training program to ensure that all staff members understand their roles and responsibilities and create a system for documentation and record-keeping.

Step Four:  Provide training and education

All staff members involved in clinical trials must be appropriately trained and educated on the importance of quality and patient safety. This includes providing training on the quality management system, relevant regulations, and the importance of patient safety. Training is essential for ensuring that everyone has the knowledge and skills needed to carry out their roles effectively. Provide regular training sessions to ensure that everyone is up to date on the latest procedures and regulations.

Step Five:  Promote effective communication

Effective communication is essential to ensure that everyone is aware of their responsibilities and can communicate openly. The training sessions, regular meetings, and feedback mechanisms are required to encourage everyone involved in the research to provide feedback on the quality culture and any issues or opportunities for improvement. This will help to identify areas that need to be addressed and provide opportunities to enhance the quality of the culture in the workplace.

Step Six:  Encourage continuous improvement

Monitor and evaluate the quality management system to identify areas for improvement. It involves conducting internal audits, analyzing quality metrics, and implementing corrective and preventive actions to address any deficiencies and continuously improve the quality management system. Recognize and reward individuals who demonstrate a commitment to quality culture. This could be in the form of bonuses, promotions, or other incentives.

Step Seven:  Demonstrate leadership commitment

Leaders within the organization should model the desired behavior by following the established policies and procedures. This will help to reinforce the importance of quality culture and inspire others to do the same. Ensure that leaders are committed to establishing and maintaining a quality culture in the workplace. Demonstrate leadership commitment by promoting and supporting a culture of quality and patient safety, providing the necessary resources and support to implement and maintain the quality management system.

Conclusion

 Incorporating a quality culture in the workplace not only increases compliance but also increases public trust in research while also advancing the field of clinical research with positive patient outcomes. If your department is looking for ways to establish or  strengthen the quality culture in the workplace, Contact us! We’d love to hear from you to discuss strategies!

The Benefits of Establishing a Quality Culture In Clinical Operations

Imagine this…you are recently hired as a Quality Associate to assist in improving inspection-readiness in the clinical operations department.  You immediately realize the following based on your observations:

  1. You are not formally introduced to the organization’s quality vision and values at your orientation session.
  2. Your mandatory training and development courses do not include or emphasize the importance of quality.
  3. The implementation of quality is rarely mentioned in the discussion of department metrics and goals.

These are warning signs of a weak quality culture within the department and minimal involvement of the quality assurance department in organization overall. 

Consequences of a weak quality culture

Quality culture in clinical research refers to an organizational mindset and set of practices that prioritize the quality and integrity of research data and the safety and well-being of study participants. Failure to establish a strong and robust quality culture in clinical research results in the following:

    • A greater risk of errors leading to serious adverse events or even death in extreme cases which can damage the reputation of the organization.

    • A trend of noncompliance with regulations which can produce poor quality data that hinders reliable conclusions from the research.

    • Poor quality control which may also result in delays and additional costs associated with correcting errors and addressing issues that arise during the research.

The benefits of establishing a quality culture

Establishing a quality culture in the workplace produces the desired clinical outcomes of clinical trials and also provides additional benefits such as:

    1. Assurance of patient safety – as clinical research involves human subjects who may be vulnerable and require protection from harm. A strong quality culture promotes adherence to strict ethical and regulatory standards, ensuring that the highest level of care and safety is provided to patients.

    1. Provision of high-quality data that is reliable and trustworthy -A quality culture ensures that data is collected, managed, and analyzed with accuracy and consistency, which enhances the credibility of the research and increases its value for future research and healthcare decision-making.

    1. Promotion of compliance to regulatory GCP requirements and standards – Compliance with these standards is necessary to gain approval for the research and to ensure that the results are acceptable for regulatory purposes.

    1. Protection of the organization’s reputation – Poor quality research can damage the reputation of the organization and undermine public trust in the research and healthcare industries.

    1. Cost reduction – Costs associated with correcting errors and addressing issues that arise during the research can be substantial. A quality culture ensures that issues are identified and addressed early on, minimizing the risk of costly mistakes and delays.

Conclusion

Establishing a quality culture promotes adherence to strict ethical and regulatory standards, ensures the collection and management of accurate and consistent data, and enhances the credibility of the research.. In addition, a quality culture helps to reduce the risk of costly mistakes and delays while protecting the organization’s reputation.  If your department is looking for ways to strengthen the quality culture in the workplace,  Contact us! We’d love to hear from you to discuss strategies!

How to have an Inspection-Ready Delegation of Authority Log

The Delegation of Authority (DOA) log is a document that records the delegation of authority for various tasks and decisions related to the conduct of a clinical trial and constitutes a critical facet of clinical trial management of participating clinical sites.  The DOA log should include the names and titles of all the members of the clinical trial team with delegated authority, as well as their respective areas of responsibility. This includes the principal investigator, sub-investigators, clinical research coordinators, and other staff involved in the conduct of the trial.

Best Practices for DOA Maintenance

Maintaining accurate and up-to-date DOA logs is critical to ensure that the trial is conducted in compliance with applicable regulations, guidelines, and standard operating procedures. Best practices for maintaining DOA logs in clinical research are as follows:

Establish clear procedures for DOA log maintenance:

Having a clear set of procedures for maintaining DOA logs is essential for ensuring consistency and accuracy. These procedures should outline who is responsible for maintaining the logs, how often they should be updated, and what information should be included.

Ensure accuracy and completeness of the information:

It is important to ensure that all information recorded in the DOA log is accurate and complete. This includes ensuring that all individuals involved in the trial are listed and that their roles and responsibilities are clearly defined.

Regularly review and update the DOA logs:

DOA logs should be reviewed and updated regularly to ensure that they remain accurate and up-to-date. This is particularly important when there are changes in personnel or when new responsibilities are assigned.

Train personnel on DOA log maintenance:

All personnel involved in the trial should be trained on the importance of maintaining accurate DOA logs and how to properly update them. This can help ensure that everyone involved in the trial understands their roles and responsibilities.

Monitor and audit DOA log maintenance:

Regular monitoring and auditing of DOA log maintenance can help ensure that procedures are being followed, information is accurate and complete, and personnel is properly trained. This can help identify and address any issues before they become more serious problems.

Conclusion

Maintaining accurate and up-to-date DOA logs is essential for effective clinical trial management at a clinical site. By following these best practices, you can help ensure that your DOA logs are accurate, complete, and secure, which can help ensure compliance with regulatory requirements and effective trial administration.

The delegation of authority (DOA) log plays a pivotal role in ensuring the efficacious and efficient conduct of clinical trials, particularly in the context of complex medical interventions. By leveraging the DOA log, clinical trial stakeholders can achieve enhanced coordination, seamless communication, and optimal resource allocation, thereby increasing your site’s audit/inspection readiness and the likelihood of successful trial outcomes.

Need further guidance or training on DOA logs/regulatory documents and how to make them audit and

inspection-ready? Contact us! We’d love to hear from you to discuss strategies!

 

6 Steps to Creating SOPs for Quality and Compliance

SOPs are crucial for ensuring compliance and conducting clinical trials. SOPs provide a standardized approach to clinical research processes, which is essential for maintaining consistency and quality across all study sites and participants. The lack of SOPs may result in several issues, such as inconsistent practices that result in disparities in data collection, analysis, and reporting, which may weaken the accuracy and dependability of the study outcomes.

Adhering to ethical and regulatory requirements in a research study can be challenging. Failure to follow standard operating procedures (SOPs) in clinical research can result in reduced efficiency and waste of time and resources in trying to determine the optimal way to perform tasks.

Why SOPs are Important for Clinical Research?

SOPs are important to clinical research as they provide:

  1. Detailed guidelines for the implementation of GCP principles which ensure that clinical trials are conducted in conformity with the ethical considerations and scientific quality standards.
  2. A standardized and streamlined approach to study procedures reducing variability of study tasks while also increasing inspection readiness strategies.
  3. Increased assurance of the safety and well-being of study participants as alignment of procedures and ethical practices and are in place.

Steps to Creating Effective SOPs

Creating effective SOPs can be challenging, but a well-designed and executed SOP can save time and resources, improve the quality of research, and reduce the risk of errors.

1.     Establish the SOP development team

The first step in creating effective SOPs for clinical research is to establish an SOP development team. The team should consist of individuals with relevant expertise and experience, including clinical research professionals, study coordinators, regulatory experts, and other stakeholders. The team should be responsible for overseeing the development and implementation of the SOPs.

2.     Identify the process

The next step is to identify the process for which the SOP is being developed. It is important to clearly define the process and the scope of the SOP. The process should be well understood by the team, and it should be clearly defined in the SOP to avoid any confusion or misinterpretation.

3.     Conduct a process mapping exercise

A process mapping exercise is a useful tool for developing SOPs for clinical research. It involves visually mapping out the process and identifying the key steps, inputs, and outputs. This exercise helps to identify areas where the process can be streamlined or improved, and it ensures that all steps are accounted for in the SOP.

4.     Develop the SOP

The next step is to develop the SOP. The SOP should be written in a clear and concise manner, using simple language. The SOP should include the purpose of the process, the step-by-step instructions for executing the process, the roles and responsibilities of team members, and any relevant references or supporting documents. The SOP should also include a section for deviations and corrective actions.

5.     Review and approve

Once the SOP has been developed, it should be reviewed and approved by the appropriate stakeholders. This includes the SOP development team, the study sponsor, the regulatory authority, and any other relevant parties. Feedback should be incorporated into the SOP, and revisions should be made as necessary.

6.     Train and implement

Training and implementation of the SOP should be conducted by the SOP development team. The team should ensure that all relevant personnel are trained on the SOP, and that the SOP is implemented consistently and effectively. The team should also monitor the implementation of the SOP and make any necessary updates or revisions.

Conclusion

In conclusion, creating effective SOPs for clinical research is critical for ensuring regulatory compliance, data integrity, and risk management. By following these six steps, the development team can create an effective and efficient SOP that will benefit the research team and the quality of the research.

 

Preparing For Pharmacovigilance FDA Inspections

The concept of pharmacovigilance—derived from the Greek and Latin ‘Pharmakon’ (medicinal substance) and Vigilia (to keep watch)—emerged in earnest among physicians and other health experts almost 200 years ago. Initially, the practice amounted primarily to letters and reports written by physicians on the safety and effectiveness of various drugs given to their patients.

Pharmacovigilance inspections (Good Pharmacovigilance Practices, GVP) are designed to assess compliance with the legally prescribed mandatory reporting of adverse drug reactions in clinical trials as well as spontaneous reports. 

The three (3) most common findings noted from FDA’s Post-marketing Adverse Drug Experience (PADE) inspections according to the Bioresearch Monitoring (BIMO) Fiscal Year 2021 Metrics  are:

  • Failure to develop written procedures for the surveillance, receipt, evaluation, and reporting of post-marketing adverse drug experiences
  • Late submission of 15-day Alert reports
  • Late submission of the annual safety report

This article will list ten (10) key areas or documentation to have ready for FDA in an upcoming GVP inspection.

What To Have Ready for an Inspection

  1. Written Procedures
    You must develop, maintain, and follow written procedures for the surveillance, receipt, evaluation, and reporting of post-marketing safety information. This includes procedures for managing safety information with contractors and business partners, as applicable.
  2. Individual Case Safety Reports (ICSRs)
    ICSRs describe one or more adverse experiences related to an individual patient or subject. A valid ICSR contains a suspect drug, an adverse experience, an identifiable patient, and an identifiable reporter.
  3. Scientific Literature Reports
    Regarding scientific literature reports, ensure that there is documented evidence of:
     Scientific literature reviews and the frequency of each review.
     Submission of expedited ICSRs for adverse experiences obtained from the published scientific and medical literature that are both serious and unexpected
    • Foreign Post-marketing Adverse Experience Reporting
      For participating affiliates, subsidiaries, contractors, and business partners outside the United States, ensure the following:
       There are written procedures that address the surveillance, receipt, evaluation, and reporting of adverse experiences.
       There is documented submission of serious and unlabeled (i.e., unexpected) adverse experiences to the FDA within 15 calendar days.
    • Solicited Safety Data
      Solicited safety data arises from organized data collection systems, which may include patient assistance programs, patient support programs, physician engagement programs, or any active solicitation of information from patients or providers, when contact between the sponsor company and the patient or provider is predictable in the context of a specific program.
    • Aggregate Safety Reports
      For each approved application or biologics license, FDA requires the submission of Periodic Reports, which describe safety information obtained during the reporting interval. The reporting interval is quarterly for the first three years following the approval of the application or license, and annually thereafter, unless FDA instructs the sponsor otherwise.
    • Contractor Oversight
      Oversight of outsourced services may include a broad range of activities to ensure that all outsourced services and activities associated with post-marketing safety are performed according to applicable FDA regulations.
    • Electronic Submissions
      Determine if safety report submissions are in an electronic format that FDA can process, review, and archive, as required.
    • Waivers
      Any post-marketing safety waivers from the regulatory requirements must follow applicable procedures and terms of the waiver.
    • Recordkeeping
      For approved drugs or biologics, ensure that all records containing information relating post-marketing safety reports (whether submitted to FDA) have been maintained for a period of 10 years, or for combination products, the longest retention period applicable.

    Conclusion

    Post-marketing safety data collection and adverse event reporting is a critical element of the Food and Drug Administration’s post-marketing safety surveillance program for FDA-regulated drug and therapeutic biologic products.  Incorporating the FDA requirements and guidance into your inspection readiness program contributes to the success of your GVP inspection.

    5 Reasons Why QA Is Essential in Clinical Trials

    The coordinated and impartial review of all clinical trial-related activities and records is known as quality assurance. In the case of a clinical study, the quality assurance department has a wide range of responsibilities. Quality Assurance (QA) departments frequently aid in inspection readiness by establishing investigator site selection guidelines and identifying service providers to be audited, such as laboratories, packaging and supply chain vendors.

    The Ongoing Challenge

    The continuous monitoring of data collection processes and data management policies at every level of the study is an ongoing challenge in managing the quality of clinical data and maintaining compliance. This includes the following:

    • verifying that the data collected during the trial is consistent with the procedure (case report form [CRF] vs. protocol)
    • ensuring the validity of the data in the CRF and data acquired in source documents (CRF vs. source documents)
    • guaranteeing that the analyzed data correspond to the CRF data (database vs. CRF)

    This confirms the need for QA involvement in clinical trials specifically in terms of inspection readiness.

    Reasons Why Quality Assurance (QA) is essential

    1. Time Saver

    While continuous monitoring during a clinical trial is a taxing task, it can save you from wasting hundreds of hours rectifying shortcomings within the trials at a later stage. Errors recognized in their initial stages are easier to modify to achieve desired outcomes. Whereas, delays can worsen the problem in clinical trials and inevitably push back the desired result, which can hinder the inspection readiness process.

    1. Money Saver

    While many believe that investing in quality assurance from the get-go is not only time-consuming and costly, it is quite the opposite. Errors during the trial stage lead to millions of dollars lost during the production stage and major delays in inspection readiness, which can further add to the cost. Sometimes dropping the trial before entering the production stage due to a lack of quality assurance become inevitable.

    1. Boosts Client Confidence

    Businesses that are known to ensure quality assurance are more likely to retain trust and confidence from clients and customers alike. During clinical trials, clients are more likely to follow the lead and trust the process when their standards of expectations align with the trials working. When boosting clients’ confidence, it is essential to highlight both the “whats” and “hows” of the trial, and quality assurance helps deliver the “hows” of the trial to keep your clients’ mind at ease.

    1. The Backbone of Consistency

    When it comes to clinical trials, it is of utmost importance that each drug produced be of the same quality to prevent ill-desired outcomes. In clinical research, the quality assurance process ensures internal consistency by scheduling regular operational checks at each level of the trial process and data collection processing to validate trial procedure compliance and data validity.

    1. Leads to Accountability

    When quality assurance is the working foundation of a clinical trial, each individual involved plays a vital role in ensuring that they deliver top-notch results in making the trial a success. Hence, the need for excellence permeates every aspect of a company in which quality assurance is at play.

    Conclusion

    Maintaining integrity and precision during a clinical trial is an ongoing, dynamic process that is the key to inspection readiness. This continuing process necessitates modifying processes and effectively conveying these adjustments to all investigators and support staff. This is why quality assurance involvement is essential and a key component to the clinical Quality Management System (cQMS) overall.

    Need to strengthen or supplement your QA component? Contact us for a free consultation! We would love to hear from you to discuss strategies!

     

    References

    • BROWN, C., 2019. Price Check: How to Cut Costs in Clinical Trials. [online] Anjusoftware.com. Available at: <https://www.anjusoftware.com/about/all-news/insights/cut-costs-clinical-trials> [Accessed 8 April 2022].
    • JLI Blog | Global Training & Education Provider. 2018. Quality Control and Quality Assurance in Clinical Trial | James Lind Blog. [online] Available at: <https://www.jliedu.com/blog/clinical-trial-quality-control-assurance/#:~:text=In%20clinical%20research%2C%20quality%20control,and%20reliability%20of%20the%20data.> [Accessed 8 April 2022].
    • Manghani, K., 2011. Quality assurance: Importance of systems and standard operating procedures. Perspectives in Clinical Research, 2(1), p.34.
    • Parashar, P., 1995. Patient Satisfaction – A valid tool of quality assurance (C. Q. I). J Family Community Med, 2(2), pp.7-8.
    • The Important Site. 2022. 10 Reasons Why Quality Assurance Is Important – The Important Site. [online] Available at: <https://theimportantsite.com/10-reasons-why-quality-assurance-is-important/#:~:text=Quality%20assurance%20is%20a%20process%20all%20organizations%20should,who%20could%20be%20with%20the%20company%20or%20independent.> [Accessed 8 April 2022].
    • Valania, M., 2006. Quality Control and Assurance in Clinical Research. [online] Applied Clinical Trials Online. Available at: <https://www.appliedclinicaltrialsonline.com/view/quality-control-and-assurance-clinical-research> [Accessed 8 April 2022].
    • WCG Avoca. n.d. Inspection Readiness: What is it and how do we get there?. [online] Available at: <https://www.theavocagroup.com/inspection-readiness-what-is-it-and-how-do-we-get-there/> [Accessed 8 April 2022].